Design and Synthesis of Novel C14-Hydroxyl Substituted Triptolide

It has long been considered that the free β hydroxyl group at C14 of triptolide (1) is essential to its potent anticancer activity. In this study, we...
0 downloads 0 Views 1MB Size
J. Med. Chem. 2009, 52, 5115–5123 5115 DOI: 10.1021/jm900342g

)

)

Design and Synthesis of Novel C14-Hydroxyl Substituted Triptolide Derivatives as Potential Selective Antitumor Agents Zheng Li,†, Zhao-Li Zhou,§, Ze-Hong Miao,‡ Li-Ping Lin,‡ Hui-Jin Feng,† Lin-Jiang Tong,‡ Jian Ding,*,§,‡ and Yuan-Chao Li*,† Department of Medicinal Chemistry and ‡Division of Anti-tumor Pharmacology, State Key Laboratory of Drug Research, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, 555 Zuchongzhi Road, Zhangjiang Hi-Tech Park, Shanghai 201203, PR China, and §Department of Pharmacology, Shenyang Pharmaceutical University, 103 Wenhua Road, Shenhe District, Shenyang 110016, PR China. These authors contributed equally. )



Received March 17, 2009

It has long been considered that the free β hydroxyl group at C14 of triptolide (1) is essential to its potent anticancer activity. In this study, we synthesized novel derivatives of 1 with a hydroxyl group substituted by epoxy groups (4-8) or a five-membered ring (11-13). Compounds (4-8) showed significant in vitro anticancer activity although less potent than 1. Although with an R oxygen configuration at the C14 position, (14S)-14,21-epoxytriptolide (4) exhibited the highest potency among all these derivatives, clearly challenging the traditional viewpoint on the necessity of C14β-hydroxyl group of compound 1. Further studies revealed that while displaying broad spectrum in vitro anticancer activity, compound 4 demonstrated prominent selective in vivo anticancer activity, particularly against human ovarian SKOV-3 and prostate PC-3 cancers with obviously lower toxicity than 1. Noticeably, compound 4 was also highly effective against multidrug resistant cancer cells. Therefore, our study gives new insights into the structure-activity relationship of 1 and also produces a promising anticancer drug candidate with unique anticancer activities.

*To whom correspondence should be addressed. For Y.C.L.: phone, þ86-21-50806600 ext 3502; fax, þ86-21-50807288; E-mail, ycli@mail. shcnc.ac.cn. For J.D.: phone, þ86-21-50806600 ext 2424; fax, þ86-2150806722; E-mail, [email protected]. a Abbreviations: TWHF, Tripterygium wilfordii Hook. f.; SRB, sulforhodamine B; MDR, multidrug resistance; VER, verapamil; ADR, adriamycin; VCR, vincristine; MTT, 3-(4,5-dimethylthiazol-2-yl)-2,5diphenyltetrazolium bromide; RTV, relative tumor volume.

some conventional chemotherapeutic drugs, 1 has similar or even superior anticancer activity, especially over p53 mutated or multidrug resistant cells.9 All of these antitumor properties mentioned above suggest that 1 should be a promising anticancer drug, however, its clinical development has been discontinued. One major obstacle for this is the severe toxicity of 1, and the tissues and organs being inflicted include gastrointestinal tracts, liver, kidney, heart, blood cells, bone marrow, testes, and ovaries as well as skin.16,17 In addition, the therapeutic window of 1 is so narrow that the toxic dose is just about twice the effective dose6 or even equal in some reports.18 Previous studies on the structure-activity relationship of 1 indicated that the characteristic hydrogen-bonded C9,C11epoxy-C14β-hydroxy system may account for its antitumor effect,19 meanwhile, the configuration of the hydroxyl group should be the β orientation.2,20 On the basis of these principles, for a long time the aim of C14 modification was just to improve the lead compound’s water solubility by carboxylation of C14β-OH to introduce water solubility enhancing moieties.21,22 However, most of these derivatives are enzymatically converted into 1 in vivo, thus retaining many of its side effects. For example, 14-succinyl triptolide sodium salt (PG490-88)23 (Figure 1), a water-soluble prodrug converted to 1 in serum, entered into phase I clinical trials in Europe for the treatment of solid tumors in 2003,23 but later this clinical trial was discontinued.24 On the other hand, some medicinal chemists have found recently that substituting C14-hydroxyl with other groups such as fluoride while keeping β orientation of the substituents could retain the cytotoxicity of 1,25 and the anticancer activity of compound 1 analogues may not be

r 2009 American Chemical Society

Published on Web 07/28/2009

Introduction a

Tripterygium wilfordii Hook. f. (TWHF ), commonly known as Lei Gong Teng (Thunder God Vine), has been used in Traditional Chinese Medicine to treat autoimmune and inflammatory diseases such as rheumatoid arthritis for centuries.1-3 Triptolide (1) (Figure 1), the major component responsible for the clinical properties of TWHF, was first isolated from TWHF extracts and characterized as a diterpenoid triepoxide lactone containing an 18 (4f3) abeo-abietane skeleton in 1972.4 Right after its isolation, 1 was shown to possess high antileukemic activity in murine models4 besides its remarkable efficacy in rheumatoid arthritis,5 and this led to extensive studies regarding its antitumor properties. Compound 1 induces growth inhibition and apoptosis against various cultured human cancer cell lines6-9 as well as on primary cultures of human tumor samples.10,11 In addition to its impressive in vivo activity against murine L1210 leukemia,4 1 could also inhibit the growth of xenografts formed by different solid tumor cells6,9,12 and the experimental metastasis of B16F10 cells.9 Meanwhile, 1 was shown to sensitize cells to Apo/Trail, tumor necrosis factor-R, or DNAdamaging chemotherapeutic agents.13-15 Compared with

pubs.acs.org/jmc

5116 Journal of Medicinal Chemistry, 2009, Vol. 52, No. 16

explained as simply as before.19 Therefore, there is still big confusion on the role of C14-hydroxyl and any derivative of 1 that retains its anticancer activity but lowers risk in toxicity is still the concern of anticancer drug development. In this study, to explore whether the C14β-hydroxyl group of 1 is faithfully critical to its anticancer activity and to find novel C14 substituted compound 1 derivatives as more promising antitumor drug candidates, we designed two series of C14-spiro-triptolide derivatives with the C14-hydroxyl substituted by a chiral epoxy group or five-membered ring, which were impossible to form intramolecular hydrogen bond and assessed their anticancer activity. Our results significantly challenge the traditional learnings and, in particular, give new insights into the structure-activity relationship of 1. Moreover, we discovered that a novel derivative of 1, compound 4, possesses unique selective in vivo anticancer activity and potential low systematic toxicity, which favorably makes it a very promising anticancer drug candidate. Chemistry The synthetic strategy followed for the preparation of the analogues 4-8 and 10-13 of compound 1 is depicted in Schemes 1 and 2. We used triptonide (3), which was extracted from TWHF of our region, as starting material and initially

Figure 1. Triptolide, epitriptolide, (14S)-14,21-epoxytriptolide and 14-succinyl triptolide sodium salt.

Li et al.

focused our attention on introducing another epoxide group at C14 position in order to obtain a series of 14,21-epoxytriptolide derivatives (Scheme 1). Quad-epoxy compound 4 was obtained from triptonide through a modified CoreyChaykovsky methylenation reaction.26 We found that treatment of triptonide with NaH and Corey methylenation reagent in anhydrous DMSO at room temperature cleanly furnished 4 in 85% yield, while in the presence of t-BuOK under the same reaction conditions provided a 61:39 mixture of 4 and its diastereoisomer 5. Nucleophilic ring-opening at the C12 position of compound 4 by 1.67% HCl in acetone provided chloride 6. Alternatively, treatment of quad-epoxy 4 with SeO2 in 1,4-dioxane under reflux introduced a hydroxyl group at C5 to give compound 7 as the main product (50% yield) and a dehydration compound 8 in 10% yield. The absolute stereochemistry of C14 in compound 4 was determined by X-ray crystallography (Figure 2). Then, we switched to the construction of the five-membered ring derivatives of 1. Treatment of triptonide with (isopropoxy-dimethylsilyl)methylmagnesium chloride followed by Tamao oxidation27 provided C14β-hydroxymethylepitriptolide 10. Modifying the two hydroxyl groups of 10 by cyclic sulfitation with thionyl chloride resulted in two diasteroisomers 11 and 12, which are different in the configuration of sulfur atom. They can both be oxidized with NaIO4 and a catalytic amount of RuCl3 3 3H2O to give sulfate 13 as the sole

Figure 2. X-ray single crystal structure of (14S)-14,21-epoxytriptolide (4).

Scheme 1. Synthesis of Derivatives 4, 5, 6, 7, and 8a

a

Reagents and conditions: (a) (CH3)3SOI, NaH, DMSO, rt, 20 min, 85% (exclusively 4); (b) (CH3)3SOI, t-BuOK, DMSO, rt, 20 min, 65%, (4/5 = 1.6:1); (c) 1.67% HCl aq, acetone, reflux 7 h, 39%; (d) SeO2, 1,4-dioxane, reflux 24 h, 60% (7/8 = 5:1).

Article

product. The absolute configuration of the sulfur atom in 11 was determined by X-ray crystallography (Figure 3). Herein, we also found another stereospecific route to synthesize compound 4 from dihydroxy 10 in two steps. Selective mesylation of the primary alcohol in 10 followed by cyclization with K2CO3 in MeOH solely afforded quad-epoxy compound 4 (Scheme 2). Results and Discussion Substitution of the C14β-Hhydroxyl Group of 1 with a Chiral Epoxy Group Retains Its in Vitro Anticancer Activity. As shown above, we obtained two series of new compound 1 analogues by substituting its C14β-hydroxyl group. To

Journal of Medicinal Chemistry, 2009, Vol. 52, No. 16

5117

examine whether the substitution affected their biological activities, we first evaluated the in vitro anticancer effects of those target compounds (4-8 and 11-13) against three human tumor cell lines derived from ovary (SK-OV-3), breast (MDA-MB-468), and prostate (PC-3) using sulforhodamine B (SRB) assays.28 The result revealed that although there is no free C14β-hydroxyl group in our new derivatives, the series of 14,21-epoxytriptolide derivatives were all effective against those three cell lines, with IC50 values ranging from 0.10 to 7.34 μM (Table 1). Among them, compound 4 exhibited the highest potency, with the lowest IC50 value (0.10 μM for SKOV-3 cells) (Table 1). Unexpectedly, compound 5, the oxygen’s direction of which was the same with that of 1, exhibited much less potency than its Table 1. In Vitro Anticancer Activity of Compound 1 Derivatives in SK-OV-3, MDA-MB-468, and PC-3 Cells IC50 (μM)a

Figure 3. X-ray single crystal structure of (14S,SS)-14-spiro14R,21-sulfinyldioxytriptolide (11).

compd

SK-OV-3

MDA-MB-468

PC-3

1 2 4 5 6 7 8 10 11 12 13

0.009 0.79 0.10 1.56 0.14 1.86 0.57 >100 >100 5.10 >100

0.01 1.32 0.18 1.92 0.21 2.37 1.14 >100 >100 >100 >100

0.02 1.60 0.27 2.70 0.47 7.34 1.80 >100 20.01 6.52 13.93

a IC50: The drug concentration required for 50% inhibition of cell proliferation, while the maximum concentration used here was 100 μM.

Scheme 2. Synthesis of Derivatives 4, 10, 11, 12, 13, and 14a

a Reagents and conditions: (a) (isopropoxy-dimethylsilyl) methyl chloride, Mg, Br-(CH2)2-Br, THF, reflux 45 min; (b) KF, KHCO3, 30% H2O2, 0 °C, 2 h, 60% over 2 steps; (c) SOCl2, Et3N, CH2Cl2, 0 °C, 2 h, 79%, (11/12 = 1.2:1); (d) NaIO4, RuCl3 3 3H2O, CH3CN/H2O = 4:1, rt, 15 min, 86%; (e) MsCl, Py, rt, 10 min, 84%; (f) K2CO3, CH3OH, rt, 20 min, 90%.

5118 Journal of Medicinal Chemistry, 2009, Vol. 52, No. 16

diastereoisomer compound 4 even though the latter possesses an oxygen atom of R direction on C14. Compound 6, which could easily transform to compound 4, showed similar but a little less cytotoxicity against those three cell lines. The data indicates that the C14β-hydroxyl group of 1 is not unchangeable even in order to generate analogues of potent anticancer activity. Our result apparently challenges the classical structure-activity relationship of 1 that considers the C14β-hydroxyl group to be essential for its anticancer activity.2,19,20 In the meantime, our result shows that introduction of a chiral epoxy group to this important site of the lead compound is an effective modification method to retain the potent anticancer activity. On the other hand, the series of analogues (11, 12, 13) with five-membered spiro sulfate or sulfite substituents on C14 nearly completely lost their cytotoxicity against MDA-MB468, and only compound 12 retained weak potency against Table 2. IC50 Calues of Compound 4 and 1 in a Panel of Human Tumor Cell Lines IC50(mean ( SD)a, μM origins blood lung stomach colon

liver kidney breast ovary prostate others

cell lines

compd 4

compd 1

Molt-4 K562 A549 SGC-7901 MKN-28 HCT-116 SW1116 HCT-15 SMMC-7721 BEL-7402 786-O MDA-MB-231 MCF-7 SK-OV-3 HO-8910 PC-3 DU-145 KB Rh30 Hela U251

0.251 ( 0.077 0.421 ( 0.125 0.702 ( 0.228 0.175 ( 0.032 2.538 ( 0.215 0.219 ( 0.037 0.907 ( 0.301 0.648 ( 0.218 0.350 ( 0.020 0.332 ( 0.069 0.386 ( 0.200 0.280 ( 0.067 0.360 ( 0.076 0.270 ( 0.023 0.538 ( 0.059 0.637 ( 0.218 0.256 ( 0.024 0.615 ( 0.120 0.293 ( 0.064 0.507 ( 0.195 0.657 ( 0.235

0.017 ( 0.002 0.050 ( 0.010 0.059 ( 0.012 0.015 ( 0.0004 0.200 ( 0.042 0.010 ( 0.003 0.052 ( 0.023 0.029 ( 0.006 0.018 ( 0.004 0.020 ( 0.005 0.022 ( 0.009 0.024 ( 0.002 0.019 ( 0.002 0.010 ( 0.001 0.028 ( 0.007 0.043 ( 0.012 0.024 ( 0.009 0.043 ( 0.019 0.014 ( 0.001 0.047 ( 0.040 0.049 ( 0.015

a IC50 (mean ( SD): Cells in 96-well plates were treated with various concentrations of compound 4 for 72 h to get the IC50 values which were determined from dose-response curves, using MTT or SRB assays; IC50 values were presented as mean ( SD of three independent experiments.

Li et al.

SKOV-3 and PC-3 cell lines. We presumed that the large space taken by C14-five-membered ring might form a spatial obstacle that prevented the interaction between these derivatives and their target molecule(s) responsible for initiating their cytotoxic effects. Compound 4 Elicits Broad-Spectrum in Vitro Antitumor Effects. To characterize the anticancer activity of compound 4, we further extended its in vitro anticancer evaluation to a panel of 21 human tumor cell lines, which come from distinct human tissues, including leukemia, lung cancer, gastric cancer, hepatoma, colon cancer, breast cancer, ovarian cancer, prostate cancer, renal cancer, cervical cancer, rhabdomyosarcoma, and glioma cells. The data showed that compound 4 exhibited a similar in vitro anticancer spectrum but reduced anticancer activities when compared to compound 1 (Table 2). Compound 4 displayed apparent cytotoxicity with IC50 values ranging from 0.18 to 0.91 μM against almost all of the above human tumor cell lines, except that this compound appeared to be relatively less effective against MKN-28 with an IC50 value of 2.54 μM. The results indicate that compound 4 possesses a broad anticancer spectrum of in vitro anticancer actions. Compound 4 Produces Direct Cytotoxic Effects on Multidrug Resistant Cancer Cell Lines. Multidrug resistance (MDR), especially to drugs of natural origin, is an important impediment to the effective chemotherapy of cancer. It has always been the focused area to develop new agents to circumvent MDR. To examine the anticancer activity of compound 4 against MDR tumor cells, we used three classical MDR cell lines (K562/A02, KB/VCR, and MCF7/ADR) that were significantly resistant to the corresponding drugs used to establish these cell lines. The IC50 values of compound 4 in these MDR cell lines were 0.74, 0.47, and 0.91 μM, while in their corresponding sensitive cell lines (K562, KB, and MCF-7), its IC50 values were 0.42, 0.61, and 0.36 μM, respectively. The results indicated that compound 4 showed approximately equivalent cytotoxicity against each MDR subline as compared with their parental cell lines, with resistance factors of 1.76, 0.77, and 2.53, respectively (Figure 4A). However, the metabolism of compound 4 was not seen via the p-glycoprotein transportation pathway because verapamil (VER), a well-characterized MDR reversal agent, did not increase its cytotoxic effect on KB/VCR cells but enhanced the cytotoxicity of VCR approximately 10-fold as expected (Figure 4B).

Figure 4. Anti-MDR activity of compound 4. (A) Resistance factors of compound 4 and reference drugs. K562/A02, KB/VCR, and MCF/ ADR cell lines are MDR cell lines derived from K562, KB, and MCF-7 cells, respectively. (B) The effect of VER on the cytotoxicity of compound 4 and VCR in KB/VCR cells. The inhibition rate was assessed by SRB assays, and each data point represents the mean of three independent experiments.

Article

Journal of Medicinal Chemistry, 2009, Vol. 52, No. 16

5119

Figure 5. Antitumor effect of compound 4 in nude mouse models. The models were generated as described in the Experimental Section. Compound 4 displayed selective in vivo antitumor activity, as indicated by its special potent effect against ovarian SK-OV-3 (A) and prostate PC-3(B) cancers but moderate action in the SGC-7901 model (C) and no effect in the SMMC-7721 model (D) and in the MDA-MB-468 model (E). No body weight difference was seen between compound 4 and vehicle treated groups and the body weight curves were illustrated in (F). Significant differences between compound 4 treatment groups and control were determined using a Student t test. * P < 0.05; ** P < 0.01.

Compound 4 Exerts Selective in Vivo Anticancer Effects. Compound 1 has been reported to have a wide in vivo anticancer spectrum with apparent effects against human liver,29 gastric,9 and breast cancers6,9 but to reveal a marginal effect against ovarian cancer in vivo.30 Here we investigated the in vivo anticancer effects of compound 4 against human breast (MDA-MB-468), liver (SMMC-7721), stomach (SGC-7901), ovary (SK-OV-3), and prostate (PC-3) cancer xenografts in nude mice. Surprisingly, the results showed that compound 4, orally administered, was highly effective against human ovarian and prostate cancer xenografts in contrast to its modest to minimal activity in the models of human gastric, hepatic, and breast cancer xenografts at the same dosage (Figure 5). The treatment with compound 4 resulted in a marked, even total, regression of tumors in the models of SK-OV-3 and PC-3 xenografts on the 21st day post the first drug administration. In the SK-OV-3 model,

compound 4 at 2 and 4 mg/kg achieved a tumor growth inhibition rate of 87.5% and 97.4%, respectively. Moreover, tumor xenografts totally disappeared in 3 mice out of 6 at 4 mg/kg and in 1 mouse out of 6 at 2 mg/kg. The similar result was seen in the PC-3 models. However, compound 4 demonstrated relatively weak antitumor effect in the model of gastric cancer SGC-7901 xenografts, with a growth inhibition rate of only 75% even at the maximal dose used (4 mg/ kg). More prominently, compound 4 led to no anticancer activity in the models of hepatic cancer SMMC-7721 and breast cancer MDA-MB-468 xenografts. These data indicate that compound 4 elicits selective anticancer effects, specifically against ovarian and prostrate cancers, although the mechanism(s) are still open to be clarified. Noticeably, compound 4 was well tolerated, not causing death or serious systematic toxicity in mice. The change in the body weight of the tested mice in the groups treated with

5120 Journal of Medicinal Chemistry, 2009, Vol. 52, No. 16

compound 4 or with vehicle was basically identical (Figure 5F). There was no distinguishable gross anatomy differences observed in livers or kidneys between the compound 4 treated groups and the vehicle groups. In contrast, the lead compound 1 was reported to cause animal death within doses 4 times9,31 or twice6 over the effective doses, and the severe toxicity and narrow therapeutic window limited its clinical development as a anticancer agent.16,17 Considering that there was a 4-time distance between the lowest effective dosage in SK-OV-3 xenografts model with T/C (%) value of 50.2 (1 mg/kg) and the highest dosage we used (4 mg/kg) in our experiments, compound 4 appears to have a larger safety window than compound 1. Detailed toxicity evaluations on compound 4 are underway that will be helpful to precisely reveal its safety at least in animals. Conclusions A series of novel derivatives of 1 as potential anticancer agents were synthesized and tested for their cytotoxicity against three human tumor cell lines. Previous studies have indicated that the characteristic hydrogen-bonded C9,C11epoxy-C14β-hydroxy system may account for the antitumor effect of compound 1 by selectively alkylating the thiol groups of key enzymes regulating tumor growth.19 But in our study, the series of 14,21-epoxytriptolide derivatives (4-8) with C14hydroxyl substituted by a chiral epoxy group which were impossible to form intramolecular hydrogen bond still retained moderate to good in vitro anticancer activity, especially compound 4, with a different oxygen direction at C14 position from that of 1, exhibited much higher potency than its diastereoisomer compound 5. On the other hand, the cytotoxicity of C14 five-membered ring substituents analogues (11, 12, 13) reduced greatly against all three cell lines. On the basis of the above results, we presumed that the size and stereo configuration of functional groups at the C14 position may exert an important influence on interaction between the derivatives of compound 1 and their target molecule(s) responsible for initiating their cytotoxic effects although the exact mechanism is still unclear. Further studies revealed that although demonstrating broad -spectrum in vitro anticancer activity, compound 4 exhibited significant selectivity in its in vivo anticancer activity, as indicated by its specially highly effectiveness against human ovarian and prostate cancers. Moreover, compound 4 showed remarkable cell killing effect on MDR tumor cells. More importantly, its larger safety window seems to make it more applicable. Therefore, this study gives new insights into the structure-activity relationship of 1 and generates a new compound (compound 4) as a promising anticancer drug candidate. Experimental Section Part of Chemistry. Mass spectra and high-resolution mass spectra were measured on a Finnigan MAT-95 mass spectrometer. Elemental analysis was performed on a Carlo Erba 1106 instrument. IR spectra were recorded on a Nicollet Magna FTIR-750 spectrometer using KBr pellets in cm-1. 1H and 13 C NMR spectra were determined on Bruker AM-300 and Bruker AM-400 instruments using tetramethylsilane as internal reference. The X-ray single crystal diffraction experiment was carried out by the Shanghai Institute of Organic Chemistry, Chinese Academy of Sciences. Silica gel 60H (200-300 mesh), manufactured by Qingdao Haiyang Chemical Group Co. (China), was used for general chromatography. HPLC analysis was used a C18 reverse phase column (Agilent Eclipse

Li et al.

XDB-C18; 5 μm; 4.6 mm  250 mm) with an Agilent 1100 series system attached to a Hewlett-Packard chromatograph manager. (14S)-14,21-Epoxytriptolide (4) and (14R)-14,21-epoxytriptolide (5). Method A. (CH3)3SOI (374 mg, 1.7 mmol) and t-BuOK (174 mg, 1.5 mmol) were dissolved in dry DMSO (6.0 mL) under Ar atmosphere. The mixture was stirred for 20 min at room temperature. An anhydrous DMSO (6.0 mL) solution of triptonide (3) (360 mg, 1.0 mmol) was added all at once to the above mixture. The mixture was stirred at room temperature until TLC analysis revealed the absence of the starting material. H2O (10.0 mL) was added to the mixture and then it was extracted with ethyl acetate. The organic layer was washed with water and brine, dried over anhydrous Na2SO4, and concentrated. The crude product was purified first by chromatography on silica gel using ethyl acetate/cyclohexane (1/5). Then we use HPLC to separate the couple of diastereomers. Finally, we gained 4 (0.149 g, 0.40 mmol, 40%) and 5 (0.093 g, 0.25 mmol, 25%) separately. Method B. (CH3)3SOI (74.8 mg, 0.34 mmol) and NaH (60%) (12 mg, 0.3 mmol) were dissolved in dry DMSO (2.5 mL) under Ar atmosphere. The mixture was stirred for 20 min at room temperature. A solution of triptonide (71.6 mg, 0.2 mmol) in dry DMSO (2 mL) was added to the above mixture. The mixture was stirred at room temperature until starting material disappeared. H2O (4.0 mL) was added to the mixture and then it was extracted with ethyl acetate. The organic layer was washed with water and brine, dried over anhydrous Na2SO4, and concentrated. The crude product was purified by chromatography on silica gel using ethyl acetate/cyclohexane (1/5) to give exclusively 4 as white solide in 85% yield (63.2 mg, 0.17 mmol). Method C. To a solution of 14 (504 mg, 1.1 mmol) in CH3OH (20.0 mL) was added K2CO3 (1.33 g, 9.6 mmol). The mixture was stirred for 20 min under room temperature. After removal of the solvent under reduced pressure, the residue was diluted with water and then extracted with ethyl acetate, washed with brine, and dried with anhydrous Na2SO4. The crude product was purified via chromatography on silica gel using ethyl acetate/cyclohexane (1/5) to give 4 in 90% yield (368 mg, 0.99 mmol). Compound 4. 1H NMR(CDCl3, 300 MHz) δ 4.67 (s, 2 H), 3.88 (d, J =2.9 Hz, 1 H), 3.52 (d, J =3.1 Hz, 1 H), 3.40 (d, J =5.5 Hz, 1 H), 2.84 (d, J =5.2 Hz, 1 H), 2.80-2.69 (m, 2 H), 2.37-2.25 (m, 1 H), 2.21-2.05 (m, 2 H), 1.91-1.80 (m, 2 H), 1.57 (dd, J =12.4, 4.9 Hz, 1 H), 1.29-1.17 (m, 1 H), 1.06 (s, 3 H), 0.86 (d, J =6.8 Hz, 3 H), 0.80 (d, J =6.8 Hz, 3 H). 13C NMR (CDCl3, 100 MHz) δ 173.2 (C), 160.2 (C), 125.2 (C), 69.9 (CH2), 65.2 (C), 65.0 (C), 58.4 (C), 56.8 (CH), 55.9 (CH), 55.6 (C), 54.1 (CH), 47.9 (CH2), 40.4 (CH), 35.6 (C), 30.2 (CH2), 23.4 (CH2), 23.2 (CH), 19.8 (CH3), 17.6 (CH3), 17.0 (CH2), 13.5 (CH3). IR (KBr) 3427, 3016, 2981, 2929, 1745, 1674, 1442, 1022 cm-1. MS (EI, 70 eV) m/z (%) 373 ([M þ 1]þ, 2), 372 (Mþ, 1), 357 (5), 343 (21), 91 (100). HRMS (EI) calcd for C21H24O6 372.1573, found 372.1578. Anal. (C21H24O6) C, H. Compound 5. 1H NMR (CDCl3, 300 MHz) δ 4.79 (dt, J = 16.8, 2.6 Hz, 1 H), 4.60 (dd, J =16.8, 2.8 Hz, 1 H), 3.81 (d, J =3.1 Hz, 1 H), 3.52 (d, J =3.1 Hz, 1 H), 3.38 (d, J =4.2 Hz, 1 H), 2.89 (d, J =4.7 Hz, 1 H), 2.85 (d, J =4.7 Hz, 1 H), 2.81-2.72 (m, 1 H), 2.39-2.27 (m, 1 H), 2.17 (dt, J =14.3, 4.2 Hz, 1 H), 2.11-1.94 (m, 1 H), 1.94-1.81 (m, 2 H), 1.47-1.29 (m, 2 H), 0.96 (s, 3 H), 0.89 (d, J =6.8 Hz, 3 H), 0.77 (d, J =6.8 Hz, 3 H). 13C NMR (CDCl3, 100 MHz) δ 173.2 (C), 161.6 (C), 125.4 (C), 70.8 (CH2), 65.9 (C), 64.6 (C), 58.3 (C), 57.2 (CH), 56.2 (CH), 55.9 (C), 54.7 (CH), 48.2 (CH2), 36.9 (CH), 36.1 (C), 28.4 (CH2), 26.9 (CH2), 23.0 (CH), 21.7 (CH3), 20.2 (CH3), 17.3 (CH3), 16.6 (CH2). IR (KBr) 3433, 2947, 2929, 1768, 1755, 1689, 1442 cm-1. MS (EI, 70 eV) m/z (%) 372 (Mþ, 1), 357 (9), 343 (18), 325 (34), 259 (100). HRMS (EI) calcd for C21H24O6 372.1573, found 372.1586. Anal. (C21H24O6) C, H. (12R,13R,14S)-12β-Chloro-13r-hydroxy-14,21-epoxytriptolide (6). To a solution of compound 4 (40 mg, 0.11 mmol) in acetone (6.0 mL) was added aq HCl (1.67%, 6.0 mL, 2.7 mmol).

Article

The mixture was gently refluxed for 7 h. After removal of most of the solvent under reduced pressure, the residue was extracted with ethyl acetate, washed with brine, and dried over anhydrous Na2SO4. The crude product was purified via chromatography on silica gel using ethyl acetate/cyclohexane (1/3) to provide 6 in 39% yield (17.5 mg, 0.043 mmol). 1H NMR (CDCl3, 300 MHz) δ 4.69 (s, 2 H), 4.16 (d, J =5.7 Hz, 1 H), 3.75 (d, J =5.7 Hz, 1 H), 3.46 (d, J =6.0 Hz, 1 H), 2.93-2.83 (m, 2 H), 2.78 (d, J =5.1 Hz, 1 H), 2.36-2.25 (m, 1 H), 2.21-2.04 (m, 2 H), 1.94-1.74 (m, 2 H), 1.59 (dd, J =12.3, 4.7 Hz, 1 H), 1.35-1.21 (m, 1 H), 1.03 (s, 3 H), 0.99 (d, J =2.2 Hz, 3 H), 0.96 (d, J =2.2 Hz, 3 H). 13C NMR (acetone-d6, 100 MHz) δ 173.7 (C), 162.2 (C), 124.6 (C), 76.6 (C), 70.7 (CH2), 69.9 (C), 60.3 (CH), 59.8 (C), 58.3 (CH), 58.2 (C), 58.2 (CH), 48.0 (CH2), 40.2 (CH), 35.8 (C), 31.3 (CH2), 28.8 (CH), 23.1 (CH2), 18.0 (CH3), 17.4 (CH2), 16.3 (CH3), 13.9 (CH3). IR (KBr) 3462, 2933, 2252, 1743, 1674, 1439, 1003 cm-1. MS (EI, 70 eV) m/z (%) 408 (Mþ, 9), 390 (3), 373 (8), 365 (100). HRMS (EI) calcd for C21H25ClO6 408.1339, found 408.1339. (5R,14S)-5r-Hydroxy-14,21-epoxytriptolide (7) and (14S)Δ5,6-dehydro-14,21-epoxytriptolide (8). To a solution of compound 4 (74.4 mg, 0.20 mmol) in 1,4-dioxane (8.0 mL) was added SeO2 (111 mg, 1.0 mmol). The mixture was gently refluxed for 24 h. Then the mixture was cooled down to room temperature, filtered through a short pad of silicon gel, and rinsed with ethyl acetate. The solvent was removed under reduced pressure. To the residue was added ethyl acetate and saturated Na2CO3. After vigorous extraction, the organic layer was washed with water and brine and dried over anhydrous Na2SO4. After concentration, the residue was purified by chromatography on silica gel using CH2Cl2 to provide 7 in 50% yield (38.8 mg, 0.1 mmol) and 8 in 10% yield (7.4 mg, 0.02 mmol). Compound 7. 1H NMR (CDCl3, 300 MHz) δ 4.92 (dt, J = 17.1, 3.1 Hz, 1 H), 4.71 (dd, J =17.1, 3.9 Hz, 1 H), 3.92 (d, J =3.3 Hz, 1 H), 3.63 (d, J =3.3 Hz,1 H), 3.40 (d, J =4.8 Hz, 1 H), 2.86 (d, J =5.1 Hz, 1 H), 2.79 (d, J =5.1 Hz, 1 H), 2.41-2.04 (m, 4 H), 1.95-1.74 (m, 2 H), 1.29 (dd, J =12.7, 4.9 Hz, 1 H), 1.12 (s, 3 H), 0.87 (d, J =6.8 Hz, 3 H), 0.84 (d, J =6.8 Hz, 3 H). 13C NMR (CDCl3, 100 MHz) δ 173.2 (C), 159.0 (C), 128.0 (C), 72.3 (C), 68.7 (CH2), 65.8 (C), 63.2 (C), 58.4 (C), 56.4 (CH), 55.5 (C), 55.0 (CH), 54.5 (CH), 48.0 (CH2), 40.6 (C), 31.0 (CH2), 24.6 (CH2), 23.4 (CH), 19.7 (CH3), 17.8 (CH3), 17.4 (CH2), 16.2 (CH3). IR (KBr) 3479, 2956, 1736, 1668, 1442, 1037 cm-1. MS (EI, 70 eV) m/z (%) 389 ([M þ 1]þ, 2), 373 (2), 359 (100), 341 (18). HRMS (EI) calcd for C21H25O7 (M þ H)þ 389.1601, found 389.1606. Anal. (C21H24O7) C, H. Compound 8. 1H NMR (CDCl3, 300 MHz) δ 6.03 (d, J =3.7 Hz, 1 H), 4.95 (d, J =16.0 Hz, 1 H), 4.83 (dd, J =16.0, 2.5 Hz, 1 H), 3.86 (d, J =3.0 Hz, 1 H), 3.56-3.48 (m, 2 H), 2.92 (d, J =4.9 Hz, 1 H), 2.86 (d, J =4.9 Hz, 1 H), 2.51-2.40 (m, 1 H), 2.362.21 (m, 1 H), 1.90 (sept, J =6.8 Hz, 1 H), 1.52-1.34 (m, 2 H), 1.29 (s, 3 H), 0.92 (d, J =6.8 Hz, 3 H), 0.78 (d, J =6.8 Hz, 3 H). 13 C NMR (CDCl3, 100 MHz) δ 173.0 (C), 153.1 (C), 140.4 (C), 126.9 (C), 121.8 (CH), 68.9 (CH2), 65.1 (C), 64.4 (C), 61.3 (C), 56.6 (CH), 55.8 (C), 54.2 (CH), 53.7 (CH), 48.4 (CH2), 37.2 (C), 30.3 (CH2), 23.1 (CH), 22.8 (CH3), 20.3 (CH3), 17.2 (CH3), 17.1 (CH2). IR (KBr) 3433, 2979, 2927, 1747, 1657, 1358, 1024 cm-1. MS (EI, 70 eV) m/z (%) 370 (Mþ, 13), 355 (22), 341 (91), 327 (95), 115 (100). HRMS (EI) calcd for C21H22O6 370.1416, found 370.1404. Anal. (C21H22O6) C, H. (14S)-14β-Hydroxymethylepitriptolide (10). Under Ar atmosphere, a portion (1.0 mL) of a solution of (isopropoxydimethylsilyl) methyl chloride (0.72 mL, 4.0 mmol) in anhydrous THF (7.0 mL) was added to Mg turnings (108 mg, 4.5 mmol). To the stirred mixture was added a few drops of 1,2-dibromoethane at 60 °C and an exothermic reaction started in several minutes. The remaining solution was added dropwise over 5 min. After the addition was completed, the gray mixture was refluxed for 45 min and then cooled to 0 °C. A solution of triptonide (3) (358 mg, 1.0 mmol) in anhydrous THF (10.0 mL) was added to the Grignard reagent (freshly prepared) at the

Journal of Medicinal Chemistry, 2009, Vol. 52, No. 16

5121

same temperature over a few minutes. After stirring at 0 °C for 2 h, the mixture was quenched with a saturated NH4Cl solution and extracted with EtOAc. The organic layer was washed with brine, dried over anhydrous Na2SO4, and concentrated under reduced pressure to give a single adduct. To a stirred mixture of colorless crude adduct, MeOH (5.0 mL), THF (8.0 mL), KHCO3 (416 mg, 4.2 mmol), and KF (464 mg, 4.9 mmol) was added H2O2 (30%, 1.1 mL, 9.71 mmol) dropwise at room temperature. The mixture was stirred at room temperature until starting material disappeared. Aqueous sat. Na2S2O3 solution (50%) was added slowly to the mixture and stirred until a negative starch/iodide test was observed. The mixture was extracted with EtOAc. The organic layer was washed with brine, dried over Na2SO4, and concentrated. The crude product was purified by chromatography on silica gel using ethyl acetate/ cyclohexane (1/3) to provide 10 in 60% yield (234 mg, 0.6 mmol). 1H NMR (CDCl3, 300 MHz) δ 4.67 (s, 2 H), 4.26 (d, J =11.8 Hz, 1 H), 3.87-3.80 (m, 2 H), 3.64 (d, J =11.5 Hz, 1 H), 3.46 (d, J =3.3 Hz, 1 H), 2.76-2.64 (m, 1 H), 2.45 (sept, J = 6.9 Hz, 1 H), 2.37-2.25 (m, 1 H), 2.23-2.04 (m, 2 H), 1.89 (t, J = 14.1 Hz, 1 H), 1.55 (dd, J =12.6, 5.2 Hz, 1 H), 1.25-1.13 (m, 1 H), 1.07 (s, 3 H), 0.91 (d, J =6.9 Hz, 3 H), 0.89 (d, J =6.9 Hz, 3 H). 13C NMR (CDCl3, 100 MHz) δ 173.2 (C), 160.2 (C), 125.4 (C), 74.4 (C), 70.0 (CH2), 67.5 (C), 65.3 (C), 65.2 (CH2), 65.0 (C), 56.5 (CH), 56.1 (CH), 54.4 (CH), 40.3 (CH), 36.0 (C), 30.1 (CH2), 25.5 (CH), 23.4 (CH2), 20.9 (CH3), 18.6 (CH3), 17.1 (CH2), 13.7 (CH3). IR (KBr) 3415, 3361, 2966, 2927, 2875, 1755, 1724, 1672, 1439, 1074, 1018 cm-1. MS (EI, 70 eV) m/z (%) 391 ([M þ 1]þ, 2), 372 (1), 71 (100). HRMS (EI) calcd for C21H27O7 (M þ H)þ 391.1757, found 391.1752. Anal. (C21H26O7) C, H. (14S,SS)-14-Spiro-14r,21-sulfinyldioxytriptolide (11) and (14S,SR)-14-Spiro-14R,21-sulfinyldioxytriptolide (12). To a solution of compound 9 (78 mg, 0.2 mmol) in anhydrous CH2Cl2 (6.0 mL) was added dry Et3N (0.21 mL, 1.6 mmol) dropwise. The mixture was then cooled to 0 °C. Under Ar atmosphere, SOCl2 (0.3 mL, 1.2 mmol) was added to the mixture carefully. After being stirred for 2 h, the mixture was quenched with water and extracted with CH2Cl2. The organic layer was washed with water and brine and dried over anhydrous Na2SO4. After removal of the solvent under reduced pressure, the crude product was purified by chromatography on silica gel using acetate/cyclohexane (1/5) to give 11 in 43% yield (39.2 mg, 0.09 mmol) and 12 in 36% yield (30.5 mg, 0.07 mmol). Compound 11. 1H NMR (CDCl3, 300 MHz) δ 4.93 (d, J =9.6 Hz, 1 H), 4.68 (s, 2 H), 4.58 (d, J =9.6 Hz, 1 H), 3.84 (d, J =3.0 Hz, 1 H), 3.71 (d, J =5.7 Hz, 1 H), 3.61 (d, J =2.9 Hz, 1 H), 2.78-2.67 (m, 1 H), 2.61 (sept, J =6.9 Hz, 1 H), 2.38-2.26 (m, 1 H), 2.25-2.05 (m, 2 H), 1.91 (t, J =14.1 Hz, 1 H), 1.54 (dd, J = 12.6, 4.5 Hz, 1 H), 1.26-1.14 (m, 1 H), 1.07 (s, 3 H), 0.95 (d, J = 6.9 Hz, 3 H), 0.93 (d, J =6.9 Hz, 3 H). 13C NMR (CDCl3, 100 MHz) δ 173.1 (C), 159.8 (C), 125.5 (C), 91.9 (C), 74.1 (CH2), 69.9 (CH2), 65.1 (C), 64.9 (C), 61.7 (C), 56.5 (CH), 55.6 (CH), 55.5 (CH), 40.4 (CH), 35.8 (C), 30.0 (CH2), 25.7 (CH), 23.3 (CH2), 20.6 (CH3), 18.7 (CH3), 17.1 (CH2), 13.5 (CH3). IR (KBr) 3475, 2972, 2933, 2875, 1745, 1680, 1441, 1219, 1018 cm-1. MS (EI, 70 eV) m/z (%) 436 (Mþ, 2), 407 (1), 393 (6), 241 (100). HRMS (EI) calcd for C21H24SO8 436.1192, found 436.1199. Compound 12. 1H NMR (CDCl3, 300 MHz) δ 4.80 (d, J =9.6 Hz, 1 H), 4.68 (s, 2 H), 4.58 (d, J =9.6 Hz, 1 H), 3.84 (d, J =3.0 Hz, 1 H), 3.79 (d, J =5.6 Hz, 1 H), 3.53 (d, J =3.0 Hz, 1 H), 2.80-2.68 (m, 1 H), 2.38-2.26 (m, 1 H), 2.24-2.02 (m, 3 H), 1.96 (t, J =14.1 Hz, 1 H), 1.54 (dd, J =12.6, 5.1 Hz, 1 H), 1.281.15 (m, 1 H), 1.10 (s, 3 H), 0.92 (d, J =6.8 Hz, 3 H), 0.90 (d, J = 6.8 Hz, 3 H). 13C NMR (CDCl3, 100 MHz) δ 173.1 (C), 159.9 (C), 125.3 (C), 92.5 (C), 74.1 (CH2), 69.9 (CH2), 66.5 (C), 65.0 (C), 61.0 (C), 58.3 (CH), 55.4 (CH), 54.7 (CH), 40.4 (CH), 35.7 (C), 30.2 (CH2), 25.7 (CH), 23.3 (CH2), 20.5 (CH3), 18.2 (CH3), 17.0 (CH2), 13.4 (CH3). IR (KBr) 3435, 2970, 2933, 2877, 2254, 1755, 1674, 1444, 1346, 1223, 1020 cm-1. MS (EI, 70 eV) m/z (%)

5122 Journal of Medicinal Chemistry, 2009, Vol. 52, No. 16

436 (Mþ, 7), 421 (9), 71 (100). HRMS (EI) calcd for C21H24SO8 436.1192, found 436.1182. (14S)-14-Spiro-14r,21-sulfonyldioxytriptolide (13). To a solution of compound 11 (40 mg, 0.092 mmol) in CH3CN (4.0 mL) was added NaIO4 (31 mg, 0.14 mmol), RuCl3 3 3H2O (6 mg, 0.028 mmol), and H2O (1.0 mL) in sequence. The mixture was stirred at room temperature for 15 min. After removal of the solvent under reduced pressure, the residue was diluted with ethyl acetate and washed with water and brine and dried over anhydrous Na2SO4. The crude product was purified by chromatography on silica gel using acetate/cyclohexane (1/4) to provide 13 in 86% yield (36 mg, 0.079 mmol). 1H NMR (CDCl3, 300 MHz) δ 4.92 (d, J = 10.0 Hz, 1 H), 4.72-4.63 (m, 3 H), 3.87 (d, J =3.0 Hz, 1 H), 3.83 (d, J =5.6 Hz, 1 H), 3.65 (d, J =3.0 Hz, 1 H), 2.79-2.68 (m, 1 H), 2.51 (sept, J =6.8 Hz, 1 H), 2.39-2.07 (m, 3 H), 1.98 (t, J =14.2 Hz, 1 H), 1.53 (dd, J = 12.6, 5.2 Hz, 1 H), 1.29-1.14 (m, 1 H), 1.09 (s, 3 H), 0.99 (d, J = 6.8 Hz, 3 H), 0.97 (d, J =6.8 Hz, 3 H). 13C NMR (CDCl3, 100 MHz) δ 172.9 (C), 159.4 (C), 125.6 (C), 91.3 (C), 73.4 (CH2), 69.9 (CH2), 66.3 (C), 65.2 (C), 61.9 (C), 58.3 (CH), 55.7 (CH), 55.5 (CH), 40.3 (CH), 35.7 (C), 30.2 (CH2), 25.5 (CH), 23.2 (CH2), 20.4 (CH3), 18.3 (CH3), 17.0 (CH2), 13.4 (CH3). IR (KBr) 3442, 2970, 2941, 1749, 1676, 1441, 1394, 1217, 1001 cm-1. MS (EI, 70 eV) m/z (%) 452 (Mþ, 8), 437 (10), 423 (10), 111 (100). HRMS (EI) calcd for C21H24SO9 452.1141, found 452.1169. (14S)-14β-(Hydroxymethyl)epitriptolide (14). Compound 10 (0.50 g, 1.3 mmol) was dissolved in dry pyridine (4.0 mL, 49.3 mmol). The solution was cooled to 0 °C, then MsCl (0.61 mL, 7.7 mmol) was added to the solution dropwise and stirred for 10 min at room temperature. After removal of the solvent under reduced pressure, the residue was diluted with water, then extracted with ethyl acetate, washed with brine, and dried with anhydrous Na2SO4. After concentration, the residue was purified by chromatography by ethyl acetate/cyclohexane (1/2) to provide 14 as colorless oil. Yield: 0.504 g (84%). 1 H NMR (CDCl3, 300 MHz) δ 4.70-4.57 (m, 4 H), 3.80 (d, J =3.2 Hz, 1 H), 3.77 (d, J =5.6 Hz, 1 H), 3.47 (d, J =3.2 Hz, 1 H), 3.10 (s, 3 H), 2.77-2.67 (m, 1 H), 2.54 (sept, J =6.9 Hz, 1 H), 2.37-2.26 (m, 1 H), 2.25-2.06 (m, 1 H), 1.91 (dd, J =14.8, 13.4 Hz, 1 H), 1.54 (dd, J =12.9, 5.1 Hz, 1 H), 1.28-1.13 (m, 2 H), 1.07 (s, 3 H), 0.92 (d, J =7.7 Hz, 3 H), 0.89 (d, J =7.7 Hz, 3 H). 13C NMR (CDCl3, 100 MHz) δ 173.2 (C), 160.1 (C), 125.4 (C), 74.5 (C), 72.0 (CH2), 69.9 (CH2), 67.8 (C), 65.1 (C), 63.3 (C), 55.9 (CH), 55.4 (CH), 54.0 (CH), 40.5 (CH), 37.5 (CH), 36.0 (C), 29.7 (CH2), 25.6 (CH3), 23.5 (CH2), 20.5 (CH3), 18.5 (CH3), 17.1 (CH2), 13.5 (CH3). IR (KBr) 3483, 3024, 2972, 2937, 1749, 1674, 1446, 1354, 1174 cm-1. MS (EI, 70 eV) m/z (%) 468 (Mþ, 1), 450 (3), 432 (1), 407 (1), 354 (17), 111 (100). HRMS (EI) calcd for C22H28SO9 468.1454, found 468.1457. Biology. Cell Lines and Cell Culture. Human gastric adenocarcinoma SGC-7901, hepatocellular carcinoma BEL-7402, renal cell carcinoma 786-O, and ovarian epitheloid carcinoma HO-8910 cell lines were obtained from the cell bank of the Shanghai Institute of Materia Medica, Chinese Academy of Sciences. Human chronic myelogenous leukemia K562, lung adenocarcinoma A549, breast cancer MCF-7, MDA-MB-231, and MDA-MB-468, colorectal cancer HCT-116 and HCT-15, oral squamous cell carcinoma KB, prostate cancer DU-145, and cervical cancer Hela cell lines were purchased from the American Type Culture Collection (Manassas, VA). Human acute lymphoblastic leukemia Molt-4, gastric adenocarcinoma MKN-28, prostate cancer PC-3, and ovarian cancer SK-OV-3 cell lines were from Japanese Foundation of Cancer Research (Tokyo, Japan). Human hepatocellular carcinoma SMMC7721 was a gift from the Second Military Medical School (Shanghai, China). The rhabdomyosarcoma cell line Rh30 was a gift from St. Jude Children’s Research Hospital (Memphis, TN). The doxorubicin-selected multidrug resistant (MDR) cell subline K562/A0232 was purchased from the Institute of Hematology, Chinese Academy of Medical Science

Li et al.

(Tianjin, China). The vincristine-selected MDR subline KB/VCR33 and doxorubicin-selected MDR subline MCF-7/ ADR34,35 was obtained from Zhongshan University of Medical Sciences (Guangzhou, China). All these cell lines except MCF-7 and MCF-7/Adr were maintained in RPMI 1640 medium (Gibco, Grand Island, NE) supplemented with 10% (v/v) heat-inactivated fetal bovine serum (Gibco), 2 mmol/L glutamine, 100 IU/mL penicillin, 100 μg/mL streptomycin, and 10 mmol/L HEPS (pH 7.4) in a humidified atmosphere of 95% air with 5% CO2 at 37 °C. MCF-7 and the MCF-7/ADR cells were grown in the same medium supplemented with 1 mmol/L sodium pyruvate and 0.01 mg/mL bovine insulin. Cytotoxicity Assays. The cytotoxicity of compound 1 derivatives and the reference drugs was examined using a panel of human tumor cell lines with the methods described before.36 Briefly, cells in 96-well plates were treated in triplicate with gradient concentrations of tested agents at 37 °C for 72 h, then assessed by the microculture tetrazolium [3-(4,5-dimethylthiazol-2-yl)-2,5-diphenyltetrazolium bromide, MTT] (Sigma, Saint Louis, MO) assay for leukemia cell lines, or by the sulforhodamin B (Sigma) assay for solid tumor cell lines. The cytotoxicity was expressed as an IC50, defined as the concentration required for 50% inhibition of cell growth compared with control cells and calculated with the logit method. Resistance factor was calculated as the ratio of IC50 in MDR cells over IC50 in the corresponding parental cells. The reference drugs ADR, VCR, and the MDR reversal agent VER were all purchased from Sigma with purity over 95%. In Vivo Antitumor Activity Assays. BALB/cA nu/nu mice aged 4-5 weeks were bred in the Shanghai Institute of Materia Medica (Shanghai, China), and all experiments were done according to the institutional ethical guidelines on animal care. Under sterile conditions, well developed tumors were cut into 1 mm3 fragments and transplanted sc into the right flank of nude mice using a trocar. When the tumor reached a volume of 100200 mm3, the mice were randomly assigned into control and treatment groups. Treatment groups receiving compound 4 (po) dissolved in vehicles and control groups were given vehicles alone. Vehicles and compound 4 were administered daily, and each experimental group contained six animals. The sizes of the tumors were measured twice a week using microcalipers. The tumor volume (V) was calculated as (length  width2)/2. The individual relative tumor volume (RTV) was calculated as follows: RTV =Vt/V0, where Vt is the measured volume each day, and V0 is the volume at the beginning of the treatment. The therapeutic effect of the compound was expressed as the volume ratio of treatment to control (T/C):37 T/C (%) =100%  (mean RTV of the treated group/mean RTV of the control group).

Acknowledgment. We thank Li-Juan Lu and Yong Xi for the animal experiments. This work was supported by a grant (no. 30721005) from the Natural Science Foundation of China (NSFC). Supporting Information Available: Elemental analysis data for 1, 2, and compounds 4, 5, 7, 8, 10, HPLC results of compounds 6, 11, 12, 13, and two crystallographic files of compounds 4 and 11 in CIF format. This material is available free of charge via the Internet at http://pubs.acs.org.

References (1) Huang, S. The Saint Peasant’s Scripture of Materia Medica (18357); Publishing House for Chinese Medicine Classics: Beijing, republished in 1982; pp 309-310. (2) Wang, X. X.; Matta, R.; Shen, G.; Nelin, L. D.; Pei, D. H.; Liu, Y. S. Mechanism of triptolide-induced apoptosis: effect on caspase activation and Bid cleavage and essentiality of the hydroxyl group of triptolide. J. Mol. Med. 2006, 84, 405–415. (3) Ding, G. S. Important Chinese herbal remedies. Clin. Ther. 1987, 9, 345–357.

Article (4) Kupchan, S. M.; Court, W. A.; Dailey, R. G., Jr.; Gilmore, C. J.; Bryan, R. F. Triptolide and Tripdiolide, Novel Antileukemic Diterpenoid Triepoxides from Tripterygium wilfordii. J. Am. Chem. Soc. 1972, 94, 7194–7195. (5) Zheng, J. R.; Fang, J. L.; Gu, K. X.; Yin, Y. Q.; Xu, L. F.; Gao, J. W.; Guo, H. Z.; Yu, Y. H.; Sun, H. Z. Screening of active antiinflammatory-immunosuppressive and antifertile components from Tripterygium wilfordii. II. Screening of 5 monomers from total glucosides of Tripterygium wilfordii (TII). Chung Kuo I. Hsueh Ko Hsueh Yuan Hsueh Pao 1987, 9, 323–328. (6) Shamon, L. A.; Pezzuto, J. M.; Graves, J. M.; Mehta, R. R.; Wangcharoentrakul, S.; Sangsuwan, R.; Chaichana, S.; Tuchinda, P.; Cleason, P.; Reutrakul, V. Evaluation of the mutagenic, cytotoxic, and antitumor potential of triptolide, a highly oxygenated diterpene isolated from Tripterygium wilfordii. Cancer Lett. 1997, 112, 113–117. (7) Yang, Y.; Liu, Z.; Tolosa, E.; Yang, J.; Li, L. Triptolide induces apoptotic death of T lymphocyte. Immunopharmacology 1998, 40, 139–149. (8) Chen, B. J. Triptolide, a novel immunosuppressive and antiinflammatory agent purified from a Chinese herb Tripterygium wilfordii Hook. f. Leuk. Lymphoma 2001, 42, 253–265. (9) Yang, S.; Chen, J.; Guo, Z.; Xu, X. M.; Wang, L.; Pei, X. F.; Yang, J.; Underhill, C. B.; Zhang, L. Triptolide inhibits the growth and metastasis of solid tumors. Mol. Cancer Ther. 2003, 2, 65–72. (10) Kiviharju, T. M.; Lecane, P. S.; Sellers, R. G.; Peehl, D. M. Antiproliferative and proapoptotic activities of triptolide (PG490), a natural product entering clinical trials, on primary cultures of human prostatic epithelial cells. Clin. Cancer Res. 2002, 8, 2666–2674. (11) Carter, B. Z.; Mak, D. H.; Schober, W. D.; McQueen, T.; Harris, D.; Estrov, Z.; Evans, R. L.; Andreeff, M. Triptolide induces caspase-dependent cell death mediated via the mitochondrial pathway in leukemic cells. Blood 2006, 108, 630–637. (12) Tengchaisri, T.; Chawengkirttikul, R.; Rachaphaew, N.; Reutrakul, V.; Sangsuwan, R.; Sirisinha, S. Antitumor activity of triptolide against cholangiocarcinoma growth in vitro and in hamsters. Cancer Lett. 1998, 133, 169–175. (13) Lee, K. Y.; Chang, W.; Qiu, D.; Kao, P. N.; Rosen, G. D. PG490 (triptolide) cooperates with tumor necrosis factor alpha to induce apoptosis in tumor cells. J. Biol. Chem. 1999, 274, 13451–13455. (14) Chang, W. T.; Kang, J. J.; Lee, K. Y.; Wei, K.; Anderson, E.; Gotmare, S.; Ross, J. A.; Rosen, G. D. Triptolide and chemotherapy cooperate in tumor cell apoptosis. A role for the p53 pathway. J. Biol. Chem. 2001, 276, 2221–2227. (15) Frese, S.; Pirnia, F.; Miescher, D.; Krajewski, S.; Borner, M. M.; Reed, J. C.; Schmid, R. A. PG490-mediated sensitization of lung cancer cells to Apo2L/TRAIL-induced apoptosis requires activation of ERK2. Oncogene 2003, 22, 5427–5435. (16) Lipsky, P. E.; Tao, X. L. A potential new treatment for rheumatoid arthritis: thunder god vine. Semin. Arthritis Rheum. 1997, 26, 713– 723. (17) Huynh, P. N.; Hikim, A. P.; Wang, C.; Stefonovic, K.; Lue, Y. H.; Leung, A.; Atienza, V.; Baravarian, S.; Reutrakul, V.; Swerdloff, R. S. Long-term effects of triptolide on spermatogenesis, epididymal sperm function, and fertility in male rats. J. Androl. 2000, 21, 689–699. (18) Zheng, J. Screening of active anti-inflammatory, immunosuppressive and antifertility components of Tripterygium wilfordii. III. A comparison of the antiinflammatory and immunosuppressive activities of 7 diterpene lactone epoxide compounds in vivo. Zhongguo Yi Xue Ke Xue Yuan Xue Bao 1991, 13, 391–397. (19) Kupchan, S. M.; Schubert, R. M. Selective Alkylation: A Biomimetic Reaction of the Antileukemic Triptolides? Science 1974, 185, 791–793.

Journal of Medicinal Chemistry, 2009, Vol. 52, No. 16

5123

(20) Yu, D. Q.; Zhang, D. M.; Wang, H. B.; Liang, X. T. Structure modification of triptolide, a diterpenoid from Tripterygium wilfordii. Acta Pharm. Sin. 1992, 27, 830–836. (21) Qi, Y. M.; Musser, J. H. Immunosuppressive compounds and methods. U.S. Patent US 5663335, 1997. (22) Dai, D.; Fidler, J. M.; Musser, J. H. Amino acid derivatives of triptolide compounds as immunemodulators and anticancer agents. U.S. Patent US 6569893, 2003. (23) Fidler, J. M.; Li, K.; Chung, C.; Wei, K.; Ross, J. A.; Gao, M.; Rosen, G. D. PG490-88, a derivative of triptolide, causes tumor regression and sensitizes tumors to chemotherapy. Mol. Cancer Ther. 2003, 2, 855–862. (24) Butler, M. S. Natural products to drugs: natural product-derived compounds in clinical trials. Nat. Prod. Rep. 2008, 25, 475–516. (25) Yutaka, A.; Yukio, H.; Tomoyo, H.; Saki, M.; Haruhiko, F.; Koichi, T.; Ritsuo, A.; Takeshi, M.; Shusuke, H. Fluorination of triptolide and its analogues and their cytotoxicity. Bioorg. Med. Chem. Lett. 2008, 18, 2459–2463. (26) Ciaccio, J. A.; Drahus, A. L.; Meis, R. M.; Tingle, C. T.; Smrtka, M.; Geneste, R. “Instant Methylide’’ Modification of the Corey-Chaykovsky Epoxide Synthesis. Synth. Commun. 2003, 33, 2135–2143. (27) Tamao, K.; Ishida, N. Silafunctional compounds in organic synthesis. 27. (Isopropoxydimethylsilyl)methyl grignard reagent: A new nucleophilic hydroxymethylating agent for aldehydes and ketones. Tetrahedron Lett. 1984, 25, 4245–4248. (28) Rubinstein, L. V.; Shoemaker, R. H.; Paull, K. D.; Simon, R. M.; Tosini, S.; Skehan, P.; Scudiero, D. A.; Monks, A.; Boyd, M. R. Comparison of in vitro anticancer-drug-screening data generated with a tetrazolium assay versus a protein assay against a diverse panel of human tumor cell lines. J. Natl. Cancer Inst. 1990, 82, 1113–1118. (29) Wu, S.; Ding, H.; Wu, S.; Tong, J.; Yin, D.; Yuan, X. Study on Anticarcinogenic acticity of Triptolide on Human Hepatocarcinoma SMMC-7721 Cells in Vitro and in Vivo. Zhong Guo Yao Shi 2007, 7, 837–839. (30) Westfall, S. D.; Nilsson, E. E.; Skinner, M. K. Role of triptolide as an adjunct chemotherapy for ovarian cancer. Chemotherapy 2008, 54, 67–76. (31) Wang, H.; Xu, J.; Wen, C.; Huang, X.; Chen, Y. Antitumor Effect of Triptolide in Vitro and in Vivo. Fujian Yi Ke Da Xue Xue Bao 2007, 41, 304–307. (32) Yang, C. Z; Luan, F. J; Xiong, D. S; Liu, B. R; Xu, Y. F; Gu, K. S. Multidrug resistance in leukemic cell line K562/A02 induced by doxorubicin. Zhongguo Yao Li Xue Bao 1995, 16, 333–337. (33) Zhang, X. H.; Zhang, F. Y.; Ji, X. J.; Li, Z. Y. Vincristine resistant human KB cell line and mechanism of multidrug resistance. Yao Xue Xue Bao 1994, 29, 246–251. (34) Fairchild, C. R.; Moscow, J. A.; O’Brien, E. E.; Cowan, K. H. Multidrug resistance in cells transfected with human genes encoding a variant P-glycoprotein and glutathione S-transferase-pi. Mol. Pharmacol. 1990, 37, 801–809. (35) Cowan, K. H.; Batist, G.; Tulpule, A.; Sinha, B. K.; Myers, C. E. Similar biochemical changes associated with multidrug resistance in human breast cancer cells and carcinogen-induced resistance to xenobiotics in rats. Proc. Natl. Acad. Sci. U.S.A 1986, 83, 9328– 9332. (36) Li, W.; Shao, Y.; Hu, L.; Zhang, X.; Chen, Y.; Tong, L.; Li, C.; Shen, X.; Ding, J. BM6, a new semisynthetic vinca alkaloid, exhibits its potent in vivo anti-tumor activities via its high binding affinity for tubulin and improved pharmacokinetic profiles. Cancer Biol. Ther. 2007, 6, 787–794. (37) Yuan, S.; Zhang, X.; Lu, L.; Xu, C.; Yang, W.; Ding, J. Anticancer activity of methoxymorpholinyl doxorubicin (PNU 152243) on human hepatocellular carcinoma. Anticancer Drugs 2004, 15, 641– 646.